1
|
Gallagher KE, Lucinde R, Bottomley C, Kaniu M, Suaad B, Mutahi M, Mwalekwa L, Ragab S, Twi-Yeboah L, Berkley JA, Hamaluba M, Karani A, Shangala J, Otiende M, Gardiner E, Mugo D, Smith PG, Tabu C, Were F, Goldblatt D, Scott JAG. Fractional Doses of Pneumococcal Conjugate Vaccine - A Noninferiority Trial. N Engl J Med 2024; 391:2003-2013. [PMID: 39330966 PMCID: PMC7616702 DOI: 10.1056/nejmoa2314620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
BACKGROUND Pneumococcal conjugate vaccines are an expensive component of the routine immunization schedule. Fractional-dose regimens may be one option to increase the sustainability of the vaccine program. METHODS We assessed whether the immunogenicity of fractional doses of the 10-valent and 13-valent pneumococcal conjugate vaccines (PCV10 [GSK] and PCV13 [Pfizer], respectively) would be noninferior to that of the full doses and analyzed the prevalence of vaccine-serotype carriage. We randomly assigned healthy infants in Kenya to one of seven equal-sized trial groups. Participants in groups A through F were assigned to receive either a fractional or full dose of PCV10 or PCV13, administered as two primary doses plus one booster dose. In group A, participants received a full dose of PCV13; group B, a 40% dose of PCV13; group C, a 20% dose of PCV13; group D, a full dose of PCV10; group E, a 40% dose of PCV10; and group F, a 20% dose of PCV10. Participants in the seventh group (group G) received a full dose of PCV10 as three primary doses without a booster. Immunogenicity was assessed 4 weeks after the primary series of doses and 4 weeks after the booster dose. Noninferiority could be declared 4 weeks after the primary series if the difference in the percentage of participants with a threshold response was not more than 10% and 4 weeks after administration of the booster if the ratio of the geometric mean concentration (GMC) of IgG was more than 0.5. A vaccine dose was prespecified as noninferior if it met the noninferiority criterion for at least 8 of the 10 vaccine types in the PCV10 groups or at least 10 of the 13 vaccine types in the PCV13 groups. Carriage was assessed when participants were 9 months and 18 months of age. RESULTS In the per-protocol analysis, 40% of a full dose of PCV13 met the noninferiority criterion for 12 of 13 serotypes after the primary series and for 13 of 13 serotypes after the booster. The immunogenicity of the 20% dose of PCV13 and of the 40% and 20% doses of PCV10 was not noninferior to that of the full doses. The prevalence of vaccine-serotype carriage was similar across the PCV13 groups at 9 months and 18 months of age. CONCLUSIONS In a three-dose schedule (two primary doses and a booster), 40% doses of PCV13 were noninferior to full doses for all included serotypes. Lower doses of PCV13 and PCV10 did not meet the criteria for noninferiority. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT03489018; Pan African Clinical Trial Registry number, PACTR202104717648755.).
Collapse
Affiliation(s)
- Katherine E Gallagher
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Ruth Lucinde
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Christian Bottomley
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mary Kaniu
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Badaud Suaad
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mary Mutahi
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Laura Mwalekwa
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Sarah Ragab
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Louise Twi-Yeboah
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - James A Berkley
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mainga Hamaluba
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Angela Karani
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Jimmy Shangala
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mark Otiende
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Elizabeth Gardiner
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Daisy Mugo
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Peter G Smith
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Collins Tabu
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Fred Were
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - David Goldblatt
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - J Anthony G Scott
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| |
Collapse
|
2
|
Tan S, Wang Y, Wei X, Xiao X, Gao L. Microneedle-mediated drug delivery for neurological diseases. Int J Pharm 2024; 661:124400. [PMID: 38950662 DOI: 10.1016/j.ijpharm.2024.124400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Neurological disorders, including brain injury, brain tumors, and neurodegenerative diseases, rank as the second leading cause of death worldwide. Exploring effective new treatments for neurological disorders has long been a hot research issue in clinical practice. Recently, microneedles (MNs) have attracted much attention due to their designation as a "painless and non-invasive" novel transdermal delivery method, characterized by their biocompatibility and sustainability. The advantages of MNs open an avenue for potential therapeutic interventions targeting neurological disorders. This review presents a concise overview of progress in the field of MNs, with highlights on the application in the treatment of neurological disorders. Notably, trends in the development of MNs and future challenges are also discussed.
Collapse
Affiliation(s)
- Shuna Tan
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Yitian Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Xuan Wei
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Xiao Xiao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Linbo Gao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
3
|
Seree-aphinan C, Rattanakaemakorn P, Suchonwanit P, Thadanipon K, Ratanapokasatit Y, Yongpisarn T, Malathum K, Simaroj P, Setthaudom C, Lohjai O, Tanrattanakorn S, Chanprapaph K. Immunogenicity of Intradermal Versus Intramuscular BNT162b2 COVID-19 Booster Vaccine in Patients with Immune-Mediated Dermatologic Diseases: A Non-Inferiority Randomized Controlled Trial. Vaccines (Basel) 2024; 12:73. [PMID: 38250886 PMCID: PMC10819288 DOI: 10.3390/vaccines12010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
The intradermal route has emerged as a dose-sparing alternative during the coronavirus disease 2019 (COVID-19) pandemic. Despite its efficacy in healthy populations, its immunogenicity has not been tested in immune-mediated dermatologic disease (IMDD) patients. This assessor-blinded, randomized-controlled, non-inferiority trial recruited patients with two representative IMDDs (i.e., psoriasis and autoimmune bullous diseases) to vaccinate with fractionated-dose intradermal (fID) or standard intramuscular (sIM) BNT162b2 vaccines as a fourth booster dose under block randomization stratified by age, sex, and their skin diseases. Post-vaccination SARS-CoV-2-specific IgG and interferon-γ responses measured 4 and 12 weeks post-intervention were serological surrogates used for demonstrating treatment effects. Mean differences in log-normalized outcome estimates were calculated with multivariable linear regression adjusting for their baseline values, systemic immunosuppressants used, and prior COVID-19 vaccination history. The non-inferiority margin was set for fID to retain >80% immunogenicity of sIM. With 109 participants included, 53 received fID (all entered an intention-to-treat analysis). The fID demonstrated non-inferiority to sIM in humoral (mean outcome estimates of sIM: 3.3, ΔfID-sIM [mean, 95%CI]: -0.1, -0.3 to 0.0) and cellular (mean outcome estimates of sIM: 3.2, ΔfID-sIM [mean, 95%CI]: 0.1, -0.2 to 0.3) immunogenicity outcomes. Two psoriasis patients from the fID arm (3.8%) developed injection-site Koebner's phenomenon. Fewer fID recipients experienced post-vaccination fever (fID vs. sIM: 1.9% vs. 12.5%, p = 0.027). The overall incidence of disease flare-ups was low without a statistically significant difference between groups. The intradermal BNT162b2 vaccine is a viable booster option for IMDD patients troubled by post-vaccination fever; its role in mitigating the risk of flare-ups remains unclear.
Collapse
Affiliation(s)
- Chutima Seree-aphinan
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
- Department of Internal Medicine, Division of Dermatology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Ploysyne Rattanakaemakorn
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
| | - Poonkiat Suchonwanit
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
| | - Kunlawat Thadanipon
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Yanisa Ratanapokasatit
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
| | - Tanat Yongpisarn
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
| | - Kumthorn Malathum
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Pornchai Simaroj
- Department of Ophthalmology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Chavachol Setthaudom
- Immunology Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.); (O.L.)
| | - Onchuma Lohjai
- Immunology Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.); (O.L.)
| | - Somsak Tanrattanakorn
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
| | - Kumutnart Chanprapaph
- Department of Medicine, Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (C.S.-a.); (P.R.); (P.S.); (Y.R.); (T.Y.); (S.T.)
| |
Collapse
|
4
|
Zhao H, Li P, Bian L, Zhang W, Jiang C, Chen Y, Kong W, Zhang Y. Immune Response of Inactivated Rabies Vaccine Inoculated via Intraperitoneal, Intramuscular, Subcutaneous and Needle-Free Injection Technology-Based Intradermal Routes in Mice. Int J Mol Sci 2023; 24:13587. [PMID: 37686393 PMCID: PMC10488038 DOI: 10.3390/ijms241713587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Inoculation routes may significantly affect vaccine performance due to the local microenvironment, antigen localization and presentation, and, therefore, final immune responses. In this study, we conducted a head-to-head comparison of immune response and safety of inactivated rabies vaccine inoculated via intraperitoneal (IP), intramuscular (IM), subcutaneous (SC) and needle-free injection technology-based intradermal (ID) routes in ICR mice. Immune response was assessed in terms of antigen-specific antibodies, antibody subtypes and neutralizing antibodies for up to 28 weeks. A live rabies virus challenge was also carried out to evaluate vaccine potency. The dynamics of inflammatory cell infiltration at the skin and muscle levels were determined via histopathological examination. The kinetics and distribution of a model antigen were also determined by using in vivo fluorescence imaging. Evidence is presented that the vaccine inoculated via the ID route resulted in the highest antigen-specific antibody and neutralizing antibody titers among all administration routes, while IP and IM routes were comparable, followed by the SC route. Antibody subtype analysis shows that the IP route elicited a Th1-biased immune response, while SC and IM administration elicited a prominent Th2-type immune response. Unexpectedly, the ID route leads to a balanced Th1 and Th2 immune response. In addition, the ID route conferred effective protection against lethal challenge with 40 LD50 of the rabies CVS strain, which was followed by IP and IM routes. Moreover, a one-third dose of the vaccine inoculated via the ID route provided comparable or higher efficacy to a full dose of the vaccine via the other three routes. The superior performance of ID inoculation over other routes is related to longer local retention at injection sites and higher lymphatic drainage. Histopathology examination reveals a transient inflammatory cell infiltration at ID and IM injection sites which peaked at 48 h and 24 h, respectively, after immunization, with all side effects disappearing within one week. These results suggest that needle-free injection technology-based ID inoculation is a promising strategy for rabies vaccination in regard to safety and efficacy.
Collapse
Affiliation(s)
- Huiting Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Peixuan Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lijun Bian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wen Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Yan Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| |
Collapse
|
5
|
Wang H, Xu J, Xiang L. Microneedle-Mediated Transcutaneous Immunization: Potential in Nucleic Acid Vaccination. Adv Healthc Mater 2023; 12:e2300339. [PMID: 37115817 DOI: 10.1002/adhm.202300339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/07/2023] [Indexed: 04/29/2023]
Abstract
Efforts aimed at exploring economical and efficient vaccination have taken center stage to combat frequent epidemics worldwide. Various vaccines have been developed for infectious diseases, among which nucleic acid vaccines have attracted much attention from researchers due to their design flexibility and wide application. However, the lack of an efficient delivery system considerably limits the clinical translation of nucleic acid vaccines. As mass vaccinations via syringes are limited by low patient compliance and high costs, microneedles (MNs), which can achieve painless, cost-effective, and efficient drug delivery, can provide an ideal vaccination strategy. The MNs can break through the stratum corneum barrier in the skin and deliver vaccines to the immune cell-rich epidermis and dermis. In addition, the feasibility of MN-mediated vaccination is demonstrated in both preclinical and clinical studies and has tremendous potential for the delivery of nucleic acid vaccines. In this work, the current status of research on MN vaccines is reviewed. Moreover, the improvements of MN-mediated nucleic acid vaccination are summarized and the challenges of its clinical translation in the future are discussed.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
6
|
Kouiavskaia D, Mirochnitchenko O, Troy S, Chumakov K. Antigenic diversity of type 1 polioviruses and its implications for the efficacy of polio vaccines. Vaccine 2023; 41:2147-2154. [PMID: 36828716 DOI: 10.1016/j.vaccine.2023.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 02/24/2023]
Abstract
Inactivated Polio Vaccines (IPV) and live Oral Polio Vaccine (OPV) were introduced in the mid-20th century, and their coordinated worldwide use led to almost complete elimination of the disease, with only one serotype of poliovirus remaining endemic in just two countries. Polio eradication will lead to discontinuation of OPV use and its replacement with IPV or other vaccines that are currently under development that will need to be tested in clinical trials. Despite decades of research, questions remain about the serological correlates of polio vaccine efficacy, specifically whether the vaccines are equally protective against immunologically different strains of the same serotype. The absence of significant morbidity does not allow use of a protection endpoint in clinical trials, so the answer could be obtained only by using surrogate markers such as immunogenicity. In this study, a panel of wild and vaccine-derived polioviruses of serotype 1 were tested in neutralization assays with sera from vaccine-immunized individuals. The results demonstrated that there was a significant difference in titers of neutralizing antibodies in human sera when measured against different strains. When measured with a homologous strain used for vaccine manufacture all subjects had detectable levels of antibodies, while neutralization tests with some heterologous strains failed to detect neutralizing antibodies in a number of subjects. Administration of a booster dose of IPV led to a significant increase in neutralizing titers against all strains. Results of the experiments using animal sera, performed to obtain more information on protectivity of neutralizing antibodies against heterologous strains, were consistent with the results obtained in the assays using human sera. These results are discussed in the context of serological biomarkers of protection against poliomyelitis, suggesting that potency of vaccines made from serologically different strains should be determined against both homologous and heterologous challenge viruses.
Collapse
Affiliation(s)
| | | | - Stephanie Troy
- Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | | |
Collapse
|
7
|
Choo JJY, McMillan CLD, Young PR, Muller DA. Microarray patches: scratching the surface of vaccine delivery. Expert Rev Vaccines 2023; 22:937-955. [PMID: 37846657 DOI: 10.1080/14760584.2023.2270598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Microneedles are emerging as a promising technology for vaccine delivery, with numerous advantages over traditional needle and syringe methods. Preclinical studies have demonstrated the effectiveness of MAPs in inducing robust immune responses over traditional needle and syringe methods, with extensive studies using vaccines targeted against different pathogens in various animal models. Critically, the clinical trials have demonstrated safety, immunogenicity, and patient acceptance for MAP-based vaccines against influenza, measles, rubella, and SARS-CoV-2. AREAS COVERED This review provides a comprehensive overview of the different types of microarray patches (MAPs) and analyses of their applications in preclinical and clinical vaccine delivery settings. This review also covers additional considerations for microneedle-based vaccination, including adjuvants that are compatible with MAPs, patient safety and factors for global vaccination campaigns. EXPERT OPINION MAP vaccine delivery can potentially be a game-changer for vaccine distribution and coverage in both high-income and low- and middle-income countries. For MAPs to reach this full potential, many critical hurdles must be overcome, such as large-scale production, regulatory compliance, and adoption by global health authorities. However, given the considerable strides made in recent years by MAP developers, it may be possible to see the first MAP-based vaccines in use within the next 5 years.
Collapse
Affiliation(s)
- Jovin J Y Choo
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Christopher L D McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - David A Muller
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Zhu T, Zhang W, Jiang P, Zhou S, Wang C, Qiu L, Shi H, Cui P, Wang J. Progress in Intradermal and Transdermal Gene Therapy with Microneedles. Pharm Res 2022; 39:2475-2486. [PMID: 36008737 DOI: 10.1007/s11095-022-03376-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/17/2022] [Indexed: 01/18/2023]
Abstract
Gene therapy is one of the most widely studied treatments and has the potential to treat a variety of intractable diseases. The skin's limited permeability, as the body's initial protective barrier, drastically inhibits the delivery effect of gene medicine. Given the potential adverse effects and physicochemical features of the medications, improving generic drug penetration into the skin barrier and achieving an effective level of target tissues remains a challenge. Microneedles have made tremendous improvements in aided gene transfer and medication delivery as a unique method. Microneedles offer the advantage of being minimally invasive and painless, as well as the ability to distribute gene medicines straight through the stratum corneum. Microneedles have been used to penetrate skin tissue with various nucleic acids and medicines in recent years, allowing for a wide range of applications in the treatment of skin ailments. This review focuses on skin-related disorders and immunity, and it primarily discusses the progress of microneedle transdermal gene therapy in recent years. It also complements the current major vectors and related microneedle gene therapy applications.
Collapse
Affiliation(s)
- Ting Zhu
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Wenya Zhang
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Pengju Jiang
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Shuwen Zhou
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Honglei Shi
- Wujin Hospital Affiliated With Jiangsu University, Changzhou, 213017, Jiangsu, People's Republic of China.
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, Jiangsu, People's Republic of China.
| | - Pengfei Cui
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China.
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, 213164, People's Republic of China.
| |
Collapse
|
9
|
Rzhevskiy A, Popov A, Pavlov C, Anissimov Y, Zvyagin A, Levin Y, Kochba E. Intradermal injection of lidocaine with a microneedle device to provide rapid local anaesthesia for peripheral intravenous cannulation: A randomised open-label placebo-controlled clinical trial. PLoS One 2022; 17:e0261641. [PMID: 35100279 PMCID: PMC8803196 DOI: 10.1371/journal.pone.0261641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 05/25/2021] [Indexed: 11/30/2022] Open
Abstract
Background Peripheral venous cannulation is one of the most common procedures in medicine. It is associated with noticeable pain and apprehension, although in most cases it is performed without any anesthesia due to lack of a painless, cost-effective option, which would provide rapid local anesthesia with subsequent significant reduction in the experienced pain. We conducted an open-label placebo-controlled clinical trial to evaluate the efficacy and safety of a 2% lidocaine injection using the commercially available microneedle device MinronJet600 (NanoPass Technologies Ltd, Israel) to achieve rapid local anesthesia prior to peripheral venous cannulation. Methods One hundred and two subjects were randomly allocated into two groups. In the first group, 100μL of lidocaine hydrochloride (2%) was injected intradermally to subjects using the MicronJet600 device in the left arm (MJ-Lido) and 100μL of saline was injected intradermally using the device in the right arm (MJ-Saline). In the second group, 100μL of lidocaine hydrochloride (2%) was injected using the MicronJet600 device into the left arm (MJ-Lido), with no injection into the right arm of subjects (No pretreatment). In both groups the intradermal injection was performed at the cannulation site prior to insertion of a 18G cannula into a median cubital vein in both arms. As a primary variable, a score of cannulation-induced pain was indicated by subjects using a 100-point visual analog scale immediately after cannulation. As a secondary variable, subjects in Group 2 also indicated their preference to receive the anaesthetic injection with MicronJet600 in the future by using the 5-point Likert scale. Also, as a secondary variable, the duration of skin numbness after lidocaine injection was indicated by performing a superficial pin-prick with a 27G needle at 15, 30 and 45 minutes, at distances of 1, 2 and 3 centimeters from the injection site. Results A significant pain reduction (11.0-fold) was achieved due to the lidocaine injection compared to the cannulation without any pretreatment (p< 0.005). After the lidocaine injection the anesthesia was effective up to 2 centimeters from the injection site and remained for up to 30 minutes. Eighty percent of subjects from the second group preferred cannulation after the lidocaine injection over cannulation without any pretreatment. No significant side effects were identified. Conclusion Intradermal injection of anaesthetic with Micronjet600 was found to be a safe and effective option for providing rapid local anesthesia for peripheral intravenous cannulation. Trial regiatration The clinical trial was registered, before the patient enrollment began, in the Research Registry publicly accessible database (registration identifier: researchregistry4662). Also, the trial was registered in ClinicalTrials.gov (registration identifier: NCT05108714) after its completion.
Collapse
Affiliation(s)
- Alexey Rzhevskiy
- Center of Biomedical Engineering, Sechenov First Moscow State Medical University, Moscow, Russia
- * E-mail:
| | - Andrei Popov
- Center of Biomedical Engineering, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Chavdar Pavlov
- Clinic of Internal Diseases Propedeutics, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Yuri Anissimov
- School of Natural Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Andrei Zvyagin
- Center of Biomedical Engineering, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | | |
Collapse
|
10
|
Ma Y, Ying Z, Li J, Gu Q, Wang X, Cai L, Shi L, Sun M. Immunogenicity of fractional-dose of inactivated poliomyelitis vaccine made from Sabin strains delivered by intradermal vaccination in Wistar rats. Biologicals 2022; 75:3-11. [DOI: 10.1016/j.biologicals.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 11/02/2022] Open
|
11
|
Microneedle-Mediated Vaccination: Innovation and Translation. Adv Drug Deliv Rev 2021; 179:113919. [PMID: 34375682 DOI: 10.1016/j.addr.2021.113919] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
Vaccine administration by subcutaneous or intramuscular injection is the most commonly prescribed route for inoculation, however, it is often associated with some deficiencies such as low compliance, high professionalism, and risk of infection. Therefore, the application of microneedles for vaccine delivery has gained widespread interests in the past few years due to its high compliance, minimal invasiveness, and convenience. This review focuses on recent advances in the development and application of microneedles for vaccination based on different delivery strategies, and introduces the current status of microneedle-mediated vaccination in clinical translation. The prospects for its application including opportunities and challenges are further discussed.
Collapse
|
12
|
Abstract
In recent years, the piezoelectric jet and atomization devices have exhibited tremendous advantages including their simple construction, and the fact that they are discreet and portable as well as low cost. They have been widely used in cell printing, spray cooling, drug delivery, and other industry fields. First, in this paper, two different concepts of jet and atomization are defined, respectively. Secondly, based on these two concepts, the piezoelectric jet and atomization devices can be divided into two different categories: piezoelectric micro jet device and piezoelectric atomization device. According to the organizational structure, piezoelectric micro jet devices can be classified into four different models: bend mode, push mode, squeeze mode, and shear mode. In addition, their development history and structural characteristics are summarized, respectively. According to the location of applied energy, there are two kinds of piezoelectric atomization devices, i.e., the static mesh atomization device and the vibration mesh atomization device, and both their advantages and drawbacks are discussed. The research achievements are summarized in three aspects of cell printing, spray cooling, and drug delivery. Finally, the future development trends of piezoelectric jet and atomization devices are prospected and forecasted.
Collapse
|
13
|
Leboux RJT, Schipper P, van Capel TMM, Kong L, van der Maaden K, Kros A, Jiskoot W, de Jong EC, Bouwstra JA. Antigen Uptake After Intradermal Microinjection Depends on Antigen Nature and Formulation, but Not on Injection Depth. FRONTIERS IN ALLERGY 2021; 2:642788. [PMID: 35386985 PMCID: PMC8974696 DOI: 10.3389/falgy.2021.642788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is an attractive alternative administration route for allergy vaccination, as the skin is rich in dendritic cells (DCs) and is easily accessible. In the skin multiple subsets of DCs with distinct roles reside at different depths. In this study antigen (=allergen for allergy) formulations were injected in ex vivo human skin in a depth-controlled manner by using a hollow microneedle injection system. Biopsies were harvested at the injection site, which were then cultured for 72 h. Subsequently, the crawled-out cells were collected from the medium and analyzed with flow cytometry. Intradermal administration of ovalbumin (OVA, model antigen) solution at various depths in the skin did not affect the migration and maturation of DCs. OVA was taken up efficiently by the DCs, and this was not affected by the injection depth. In contrast, Bet v 1, the major allergen in birch pollen allergy, was barely taken up by dermal DCs (dDCs). Antigens were more efficiently taken up by CD14+ dDCs than CD1a+ dDCs, which in turn were more efficient at taken up antigen than Langerhans cells. Subsequently, both OVA and Bet v 1 were formulated in cationic and anionic liposomes, which altered antigen uptake drastically following intradermal microinjection. While OVA uptake was reduced by formulation in liposomes, Bet v 1 uptake in dDCs was increased by encapsulation in both cationic and anionic liposomes. This highlights the potential use of liposomes as adjuvant in intradermal allergy vaccine delivery. In conclusion, we observed that antigen uptake after intradermal injection was not affected by injection depth, but varied between different antigens and formulation.
Collapse
Affiliation(s)
- Romain J. T. Leboux
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Pim Schipper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Toni M. M. van Capel
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | - Lily Kong
- Division of Supramolecular Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
- Tongji School of Pharmacy, HuaZhong University of Science and Technology, Wuhan, China
| | - Koen van der Maaden
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- TECO Development GmbH, Rheinbach, Germany
| | - Alexander Kros
- Division of Supramolecular Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
- Esther C. de Jong
| | - Joke A. Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
- *Correspondence: Joke A. Bouwstra
| |
Collapse
|
14
|
Korkmaz E, Balmert SC, Sumpter TL, Carey CD, Erdos G, Falo LD. Microarray patches enable the development of skin-targeted vaccines against COVID-19. Adv Drug Deliv Rev 2021; 171:164-186. [PMID: 33539853 PMCID: PMC8060128 DOI: 10.1016/j.addr.2021.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is a serious threat to global health and the global economy. The ongoing race to develop a safe and efficacious vaccine to prevent infection by SARS-CoV-2, the causative agent for COVID-19, highlights the importance of vaccination to combat infectious pathogens. The highly accessible cutaneous microenvironment is an ideal target for vaccination since the skin harbors a high density of antigen-presenting cells and immune accessory cells with broad innate immune functions. Microarray patches (MAPs) are an attractive intracutaneous biocargo delivery system that enables safe, reproducible, and controlled administration of vaccine components (antigens, with or without adjuvants) to defined skin microenvironments. This review describes the structure of the SARS-CoV-2 virus and relevant antigenic targets for vaccination, summarizes key concepts of skin immunobiology in the context of prophylactic immunization, and presents an overview of MAP-mediated cutaneous vaccine delivery. Concluding remarks on MAP-based skin immunization are provided to contribute to the rational development of safe and effective MAP-delivered vaccines against emerging infectious diseases, including COVID-19.
Collapse
Affiliation(s)
- Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Stephen C Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
15
|
Schnyder JL, Garcia Garrido HM, De Pijper CA, Daams JG, Stijnis C, Goorhuis A, Grobusch MP. Comparison of equivalent fractional vaccine doses delivered by intradermal and intramuscular or subcutaneous routes: A systematic review. Travel Med Infect Dis 2021; 41:102007. [PMID: 33711425 DOI: 10.1016/j.tmaid.2021.102007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND For certain vaccines, dosing can be reduced by intradermal (ID) immunization without loss of immunogenicity, as an alternative to standard routes of administration. However, a certain level of dose-sparing might also be achieved by reducing doses of intramuscular (IM) or subcutaneous (SC) vaccines. METHOD We conducted a systematic review comparing identical reduced amounts of antigen delivered by either ID, or IM/SC routes (PROSPERO registration no. CRD42020151725). RESULTS Of 6015 articles identified, we included 26 articles, covering eight different vaccines. Equivalent immune responses were demonstrated in 19/26 studies, and 7/26 studies suggested inferior immune responses after IM/SC immunization. CONCLUSIONS We conclude that fractional dosed IM/SC vaccination is at best as immunogenic, but potentially inferior to ID vaccination. The safety profiles were at large comparable, although minor local adverse events were more common after ID delivery. Future vaccine trials, depending on the platform used, should add a fractional dose IM/SC arm, besides a fractional dose ID arm.
Collapse
Affiliation(s)
- Jenny L Schnyder
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Hannah M Garcia Garrido
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Cornelis A De Pijper
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Joost G Daams
- Medical Library, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Cornelis Stijnis
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Abraham Goorhuis
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Martin P Grobusch
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Singh RK, Malosse C, Davies J, Malissen B, Kochba E, Levin Y, Birchall JC, Coulman SA, Mous J, McAteer MA, Dayan CM, Henri S, Wong FS. Using gold nanoparticles for enhanced intradermal delivery of poorly soluble auto-antigenic peptides. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 32:102321. [PMID: 33184020 DOI: 10.1016/j.nano.2020.102321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/08/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022]
Abstract
Ultra-small 1-2 nm gold nanoparticles (NP) were conjugated with a poorly-soluble peptide auto-antigen, associated with type 1 diabetes, to modify the peptide pharmacokinetics, following its intradermal delivery. Peptide distribution was characterized, in vivo, after delivery using either conventional intradermal injection or a hollow microneedle device. The poorly-soluble peptide was effectively presented in distant lymph nodes (LN), spleen and draining LN when conjugated to the nanoparticles, whereas peptide alone was only presented in the draining LN. By contrast, nanoparticle conjugation to a highly-soluble peptide did not enhance in vivo distribution. Transfer of both free peptide and peptide-NPs from the skin to LN was reduced in mice lacking lymphoid homing receptor CCR7, suggesting that both are actively transported by migrating dendritic cells to LN. Collectively, these data demonstrate that intradermally administered ultra-small gold nanoparticles can widen the distribution of poorly-soluble auto-antigenic peptides to multiple lymphoid organs, thus enhancing their use as potential therapeutics.
Collapse
Affiliation(s)
- Ravinder K Singh
- Division of Infection & Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | - Camille Malosse
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Joanne Davies
- Division of Infection & Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France; Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | | | - Yotam Levin
- NanoPass Technologies Ltd., Nes Ziona, Israel
| | - James C Birchall
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, UK
| | - Sion A Coulman
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, UK
| | - Jan Mous
- Midatech Pharma PLC, Cardiff, UK
| | | | - Colin M Dayan
- Division of Infection & Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK.
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - F Susan Wong
- Division of Infection & Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| |
Collapse
|
17
|
Hossain MK, Ahmed T, Bhusal P, Subedi RK, Salahshoori I, Soltani M, Hassanzadeganroudsari M. Microneedle Systems for Vaccine Delivery: the story so far. Expert Rev Vaccines 2021; 19:1153-1166. [PMID: 33427523 DOI: 10.1080/14760584.2020.1874928] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Vaccine delivery via a microneedle (MN) system has been identified as a potential alternative to conventional vaccine delivery. MN can be self-administered, is pain-free and is capable of producing superior immunogenicity. Over the last few decades, significant research has been carried out in this area, and this review aims to provide a comprehensive picture on the progress of this delivery platform. AREAS COVERED This review highlights the potential role of skin as a vaccine delivery route using a microneedle system, examines recent advancements in microneedle fabrication techniques, and provides an update on potential preclinical and clinical studies on vaccine delivery through microneedle systems against various infectious diseases. Articles for the review study were searched electronically in PubMed, Google, Google Scholar, and Science Direct using specific keywords to cover the scope of the article. The advanced search strategy was employed to identify the most relevant articles. EXPERT OPINION A significant number of MN mediated vaccine candidates have shown promising results in preclinical and clinical trials. The recent emergence of cleanroom free, 3D or additive manufacturing of MN systems and stability, together with the dose-sparing capacity of the Nanopatch® system, have made this platform, commercially, highly lucrative.
Collapse
Affiliation(s)
- Md Kamal Hossain
- Institute for Health and Sport, Victoria University , Melbourne, VIC, Australia
| | - Taksim Ahmed
- Leslie Dan Faculty of Pharmacy, University of Toronto , Toronto, Ontario, Canada
| | - Prabhat Bhusal
- School of Pharmacy, University of Otago , Dunedin New Zealand
| | | | - Iman Salahshoori
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University , Tehran, Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology , Tehran, Iran.,Department of Electrical and Computer Engineering, Faculty of Engineering, School of Optometry and Vision Science, Faculty of Science, University of Waterloo , Waterloo, Ontario, Canada.,Centre for Biotechnology and Bioengineering (CBB), University of Waterloo , Waterloo, Ontario, Canada.,Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology , Tehran, Iran
| | - Majid Hassanzadeganroudsari
- Institute for Health and Sport, Victoria University , Melbourne, VIC, Australia.,Department of Chemical Engineering, Science and Research Branch, Islamic Azad University , Tehran, Iran
| |
Collapse
|
18
|
Amani H, Shahbazi MA, D'Amico C, Fontana F, Abbaszadeh S, Santos HA. Microneedles for painless transdermal immunotherapeutic applications. J Control Release 2020; 330:185-217. [PMID: 33340568 DOI: 10.1016/j.jconrel.2020.12.019] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022]
Abstract
Immunotherapy has recently garnered plenty of attention to improve the clinical outcomes in the treatment of various diseases. However, owing to the dynamic nature of the immune system, this approach has often been challenged by concerns regarding the lack of adequate long-term responses in patients. The development of microneedles (MNs) has resulted in the improvement and expansion of immuno-reprogramming strategies due to the housing of high accumulation of dendritic cells, macrophages, lymphocytes, and mast cells in the dermis layer of the skin. In addition, MNs possess many outstanding properties, such as the ability for the painless traverse of the stratum corneum, minimal invasiveness, facile fabrication, excellent biocompatibility, convenient administration, and bypassing the first pass metabolism that allows direct translocation of therapeutics into the systematic circulation. These advantages make MNs excellent candidates for the delivery of immunological biomolecules to the dermal antigen-presenting cells in the skin with the aim of vaccinating or treating different diseases, such as cancer and autoimmune disorders, with minimal invasiveness and side effects. This review discusses the recent advances in engineered MNs and tackles limitations relevant to traditional immunotherapy of various hard-to-treat diseases.
Collapse
Affiliation(s)
- Hamed Amani
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran.
| | - Carmine D'Amico
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Samin Abbaszadeh
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran; Department of Pharmacology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
19
|
The Current Status of Clinical Research Involving Microneedles: A Systematic Review. Pharmaceutics 2020; 12:pharmaceutics12111113. [PMID: 33228098 PMCID: PMC7699365 DOI: 10.3390/pharmaceutics12111113] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/05/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, a number of clinical trials have been published on the efficacy and safety of drug delivery using microneedles (MNs). This review aims to systematically summarize and analyze the current evidence including the clinical effect and safety of MNs. Three electronic databases, including PubMed, were used to search the literature for randomized controlled trials (RCTs) and clinical controlled trials (CCTs) that evaluated the therapeutic efficacy of MNs from their inception to 28 June 2018. Data were extracted according to the characteristics of study subjects; disorder, types, and details of the intervention (MNs) and control groups; outcome measurements; effectiveness; and incidence of adverse events (AEs). Overall, 31 RCTs and seven CCTs met the inclusion criteria. Although MNs were commonly used in skin-related studies, evaluating the effects of MNs was difficult because many studies did not provide adequate comparison values between groups. For osteoporosis treatment, vaccine, and insulin delivery studies, MNs were comparable to or more effective than the gold standard. Regarding the safety of MNs, most AEs reported in each study were minor (grade 1 or 2). A well-designed RCT is necessary to clearly evaluate the effectiveness of MNs in the future.
Collapse
|
20
|
Xie L, Zeng H, Sun J, Qian W. Engineering Microneedles for Therapy and Diagnosis: A Survey. MICROMACHINES 2020; 11:E271. [PMID: 32150866 PMCID: PMC7143426 DOI: 10.3390/mi11030271] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Microneedle (MN) technology is a rising star in the point-of-care (POC) field, which has gained increasing attention from scientists and clinics. MN-based POC devices show great potential for detecting various analytes of clinical interests and transdermal drug delivery in a minimally invasive manner owing to MNs' micro-size sharp tips and ease of use. This review aims to go through the recent achievements in MN-based devices by investigating the selection of materials, fabrication techniques, classification, and application, respectively. We further highlight critical aspects of MN platforms for transdermal biofluids extraction, diagnosis, and drug delivery assisted disease therapy. Moreover, multifunctional MNs for stimulus-responsive drug delivery systems were discussed, which show incredible potential for accurate and efficient disease treatment in dynamic environments for a long period of time. In addition, we also discuss the remaining challenges and emerging trend of MN-based POC devices from the bench to the bedside.
Collapse
Affiliation(s)
- Liping Xie
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China;
| | - Hedele Zeng
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China;
| | - Jianjun Sun
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Wei Qian
- Department of Electrical and Computer Engineering, University of Texas, EI Paso, TX 79968, USA;
| |
Collapse
|
21
|
Badizadegan K, Goodson JL, Rota PA, Thompson KM. The potential role of using vaccine patches to induce immunity: platform and pathways to innovation and commercialization. Expert Rev Vaccines 2020; 19:175-194. [PMID: 32182145 PMCID: PMC7814398 DOI: 10.1080/14760584.2020.1732215] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/12/2020] [Indexed: 01/14/2023]
Abstract
Introduction: In the last two decades, the evidence related to using vaccine patches with multiple short projections (≤1 mm) to deliver vaccines through the skin increased significantly and demonstrated their potential as an innovative delivery platform.Areas covered: We review the vaccine patch literature published in English as of 1 March 2019, as well as available information from key stakeholders related to vaccine patches as a platform. We identify key research topics related to basic and translational science on skin physical properties and immunobiology, patch development, and vaccine manufacturing.Expert opinion: Currently, vaccine patch developers continue to address some basic science and other platform issues in the context of developing a potential vaccine patch presentation for an existing or new vaccine. Additional clinical data and manufacturing experience could shift the balance toward incentivizing existing vaccine manufactures to further explore the use of vaccine patches to deliver their products. Incentives for innovation of vaccine patches differ for developed and developing countries, which will necessitate different strategies (e.g. public-private partnerships, push, or pull mechanisms) to support the basic and applied research needed to ensure a strong evidence base and to overcome translational barriers for vaccine patches as a delivery platform.
Collapse
Affiliation(s)
| | - James L Goodson
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul A Rota
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | |
Collapse
|
22
|
Abstract
Under many circumstances, prophylactic immunizations are considered as the only possible strategy to control infectious diseases. Considerable efforts are typically invested in immunogen selection but, erroneously, the route of administration is not usually a major concern despite the fact that it can strongly influence efficacy. The skin is now considered a key component of the lymphatic system with tremendous potential as a target for vaccination. The purpose of this review is to present the immunological basis of the skin-associated lymphoid tissue, so as to provide understanding of the skin vaccination strategies. Several strategies are currently being developed for the transcutaneous delivery of antigens. The classical, mechanical or chemical disruptions versus the newest approaches based on microneedles for antigen delivery through the skin are discussed herein.
Collapse
|
23
|
Jaiswal N, Singh S, Agarwal A, Chauhan A, Thumburu KK, Kaur H, Singh M. Equivalent schedules of intradermal fractional dose versus intramuscular full dose of inactivated polio vaccine for prevention of poliomyelitis. Cochrane Database Syst Rev 2019; 12:CD011780. [PMID: 31858595 PMCID: PMC6923520 DOI: 10.1002/14651858.cd011780.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Poliomyelitis is a debilitating and deadly infection. Despite exponential growth in medical science, there is still no cure for the disease, which is caused by three types of wild polioviruses: types 1, 2, and 3. According to the Global Polio Eradication Initiative (GPEI), wild poliovirus is still in circulation in three countries, and fresh cases have been reported even in the year 2018. Due to the administration of live vaccines, the risk for vaccine-derived poliovirus (VDPV) is high in areas that are free from wild polioviruses. This is evident based on the fact that VDPV caused 20 outbreaks between 2000 and 2011. Recent recommendations from the World Health Organization favoured the inclusion of inactivated poliovirus vaccine (IPV) in the global immunisation schedule. IPV can be delivered in two ways: intramuscularly and intradermally. IPV was previously administered intramuscularly, but shortages in vaccine supplies, coupled with the higher costs of the vaccines, led to the innovation of delivering a fractional dose (one-fifth) of IPV intradermally. However, there is uncertainty regarding the efficacy, immunogenicity, and safety of an intradermal, fractional dose of IPV compared to an intramuscular, full dose of IPV. OBJECTIVES To compare the immunogenicity and efficacy of an inactivated poliovirus vaccine (IPV) in equivalent immunisation schedules using fractional-dose IPV given via the intradermal route versus full-dose IPV given via the intramuscular route. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, 10 other databases, and two trial registers up to February 2019. We also searched the GPEI website and scanned the bibliographies of key studies and reviews in order to identify any additional published and unpublished trials in this area not captured by our electronic searches. SELECTION CRITERIA Randomised controlled trials (RCTs) and quasi-RCTs of healthy individuals of any age who are eligible for immunisation with IPV, comparing intradermal fractional-dose (one-fifth) IPV to intramuscular full-dose IPV. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 13 RCTs involving a total of 7292 participants, both children (n = 6402) and adults (n = 890). Nine studies were conducted in middle-income countries, three studies in high-income countries, and only one study in a low-income country. Five studies did not report methods of randomisation, and one study failed to conceal the allocations. Eleven studies did not blind participants, and six studies did not blind outcome assessments. Two studies had high attrition rates, and one study selectively reported the results. Three studies were funded by pharmaceutical companies. Paralytic poliomyelitis. No study reported data on this outcome. Seroconversion rates. These were significantly higher for all three types of wild poliovirus for children given intramuscular full-dose IPV after a single primary dose and two primary doses, but only significantly higher for type two wild poliovirus given intramuscularly after three primary doses: • dose one (six studies): poliovirus type 1 (odds ratio (OR) 0.30, 95% confidence interval (CI) 0.22 to 0.41; 2570 children); poliovirus type 2 (OR 0.43, 95% CI 0.31 to 0.60; 2567 children); poliovirus type 3 (OR 0.19, 95% CI 0.12 to 0.30; 2571 children); • dose two (three studies): poliovirus type 1 (OR 0.23, 95% CI 0.16 to 0.33; 981 children); poliovirus type 2 (OR 0.41, 95% CI 0.28 to 0.60; 853 children); and poliovirus type 3 (OR 0.12, 95% CI 0.07 to 0.22; 855 children); and • dose three (three studies): poliovirus type 1 (OR 0.45, 95% CI 0.07 to 3.15; 973 children); poliovirus type 2 (OR 0.34, 95% CI 0.19 to 0.63; 973 children); and poliovirus type 3 (OR 0.18, 95% CI 0.01 to 2.58; 973 children). Using the GRADE approach, we rated the certainty of the evidence as low or very low for seroconversion rate (after a single, two, or three primary doses) for all three poliovirus types due to significant risk of bias, heterogeneity, and indirectness in applicability/generalisability. Geometric mean titres. No study reported mean antibody titres. Median antibody titres were higher for intramuscular full-dose IPV (7 studies with 4887 children); although these studies also reported a rise in antibody titres in the intradermal group, none reported the duration for which the titres remained high. Any vaccine-related adverse event. Five studies (2217 children) reported more adverse events, such as fever and redness, in the intradermal group, whilst two studies (1904 children) reported more adverse events in the intramuscular group. AUTHORS' CONCLUSIONS There is low- and very low-certainty evidence that intramuscular full-dose IPV may result in a slight increase in seroconversion rates for all three types of wild poliovirus, compared with intradermal fractional-dose IPV. We are uncertain whether intradermal fractional-dose (one-fifth) IPV has better protective effects and causes fewer adverse events in children than intramuscular full-dose IPV.
Collapse
Affiliation(s)
- Nishant Jaiswal
- Postgraduate Institute of Medical Education and ResearchICMR Advanced Centre for Evidence‐Based Child HealthSector 12ChandigarhIndia160012
| | - Shreya Singh
- Postgraduate Institute of Medical Education and ResearchDepartment of Medical MicrobiologyResearch Block A, Sector 12ChandigarhChandigarhIndia160012
| | - Amit Agarwal
- Postgraduate Institute of Medical Education and ResearchICMR Advanced Centre for Evidence‐Based Child HealthSector 12ChandigarhIndia160012
| | - Anil Chauhan
- Postgraduate Institute of Medical Education and ResearchICMR Advanced Centre for Evidence‐Based Child HealthSector 12ChandigarhIndia160012
| | - Kiran K Thumburu
- Postgraduate Institute of Medical Education and ResearchICMR Advanced Centre for Evidence‐Based Child HealthSector 12ChandigarhIndia160012
| | - Harpreet Kaur
- Panjab UniversityUniversity Business SchoolSector 14ChandigarhIndia160014
| | - Meenu Singh
- Postgraduate Institute of Medical Education and ResearchDepartment of PediatricsSector 12ChandigarhIndia160012
| | | |
Collapse
|
24
|
Universal ELISA for quantification of D-antigen in inactivated poliovirus vaccines. J Virol Methods 2019; 276:113785. [PMID: 31765719 DOI: 10.1016/j.jviromet.2019.113785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 11/20/2022]
Abstract
To address the biosafety and biosecurity concerns related to the manufacture of inactivated polio vaccine (IPV), several manufacturers started producing it from attenuated Sabin strains. Slight immunological differences between wild and attenuated strains create a challenge for testing IPV potency, which is defined as the content of protective D-antigen determined in an ELISA test. Some ELISA reagents selected for testing conventional IPV made from wild strains (cIPV) may not be suitable for testing Sabin IPV (sIPV). This paper describes an ELISA procedure using human monoclonal antibodies selected to capture equally well both wild and attenuated strains of poliovirus. A unique monoclonal antibody neutralizing all three serotypes of poliovirus was used as the detection antibody. The method was shown to detect only D-antigen of both conventional and Sabin IPV and to be strictly serotype-specific. The method is highly sensitive and robust and produces linear results in a wide range of concentrations. We have also found that reference standards used for measuring potency of cIPV and sIPV must be made from respective vaccines. This makes it impossible to cross-calibrate potency reagents made from heterologous vaccine and requires the establishment of a new unit to measure potency of sIPV that is different from conventional D-antigen unit.
Collapse
|
25
|
Pires LR, Vinayakumar KB, Turos M, Miguel V, Gaspar J. A Perspective on Microneedle-Based Drug Delivery and Diagnostics in Paediatrics. J Pers Med 2019; 9:jpm9040049. [PMID: 31731656 PMCID: PMC6963643 DOI: 10.3390/jpm9040049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
Microneedles (MNs) have been extensively explored in the literature as a means to deliver drugs in the skin, surpassing the stratum corneum permeability barrier. MNs are potentially easy to produce and may allow the self-administration of drugs without causing pain or bleeding. More recently, MNs have been investigated to collect/assess the interstitial fluid in order to monitor or detect specific biomarkers. The integration of these two concepts in closed-loop devices holds the promise of automated and minimally invasive disease detection/monitoring and therapy. These assure low invasiveness and, importantly, open a window of opportunity for the application of population-specific and personalised therapies.
Collapse
Affiliation(s)
- Liliana R Pires
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (L.R.P.); (J.G.)
| | - KB Vinayakumar
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (L.R.P.); (J.G.)
- Correspondence: or
| | - Maria Turos
- University of Oviedo, 33006 Asturias, Spain;
| | - Verónica Miguel
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa”, 28049 Madrid, Spain;
| | - João Gaspar
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (L.R.P.); (J.G.)
| |
Collapse
|
26
|
Chang L, Lim BCW, Flaherty GT, Torresi J. Travel vaccination recommendations and infection risk in HIV-positive travellers. J Travel Med 2019; 26:5486056. [PMID: 31066446 DOI: 10.1093/jtm/taz034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND With the advent of highly active antiretroviral drugs for the treatment of human immunodeficiency virus (HIV) it has become possible for people with HIV to travel to destinations that may place them at risk of a number of infectious diseases. Prevention of infections by vaccination is therefore of paramount importance for these travellers. However, vaccine responsiveness in HIV-positive individuals is not infrequently reduced compared to HIV-negative individuals. An understanding of the expected immune responses to vaccines in HIV-positive travellers is therefore important in planning the best approach to a pretravel consultation. METHODS A PubMed search was performed on HIV or acquired immune deficiency syndrome together with a search for specific vaccines. Review of the literature was performed to develop recommendations on vaccinations for HIV-positive travellers to high-risk destinations. RESULTS The immune responses to several vaccines are reduced in HIV-positive people. In the case of vaccines for hepatitis A, hepatitis B, influenza, pneumococcus, meningococcus and yellow fever there is a good body of data in the literature showing reduced immune responsiveness and also to help guide appropriate vaccination strategies. For other vaccines like Japanese encephalitis, rabies, typhoid fever, polio and cholera the data are not as robust; however, it is still possible to gain some understanding of the reduced responses seen with these vaccines. CONCLUSION This review provides a summary of the immunological responses to commonly used vaccines for the HIV-positive travellers. This information will help guide travel medicine practitioners in making decisions about vaccination and boosting of travellers with HIV.
Collapse
Affiliation(s)
- Lisa Chang
- Department of Microbiology, Dorevitch Pathology, Melbourne, Victoria, Australia
| | - Bryan Chang Wei Lim
- School of Medicine, National University of Ireland, Galway, Ireland.,School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Gerard T Flaherty
- School of Medicine, National University of Ireland, Galway, Ireland.,School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Joseph Torresi
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
27
|
Donadei A, Kraan H, Ophorst O, Flynn O, O'Mahony C, Soema PC, Moore AC. Skin delivery of trivalent Sabin inactivated poliovirus vaccine using dissolvable microneedle patches induces neutralizing antibodies. J Control Release 2019; 311-312:96-103. [PMID: 31484041 DOI: 10.1016/j.jconrel.2019.08.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/27/2019] [Accepted: 08/31/2019] [Indexed: 12/30/2022]
Abstract
The cessation of the oral poliovirus vaccine (OPV) and the inclusion of inactivated poliovirus (IPV) into all routine immunization programmes, strengthens the need for new IPV options. Several novel delivery technologies are being assessed that permit simple yet efficacious and potentially dose-sparing administration of IPV. Current disadvantages of conventional liquid IPV include the dependence on cold chain and the need for injection, resulting in high costs, production of hazardous sharps waste and requiring sufficiently trained personnel. In the current study, a dissolvable microneedle (DMN) patch for skin administration that incorporates trivalent inactivated Sabin poliovirus vaccine (sIPV) was developed. Microneedles were physically stable in the ambient environment for at least 30 min and efficiently penetrated skin. Polio-specific IgG antibodies that were able to neutralize the virus were induced in rats upon administration using trivalent sIPV-containing microneedle patches. These sIPV-patch-induced neutralizing antibody responses were comparable to higher vaccine doses delivered intramuscularly for type 1 and type 3 poliovirus serotypes. Moreover, applying the patches to the flank elicited a significantly higher antibody response compared to their administration to the ear. This study progresses the development of a skin patch-based technology that would simplify vaccine administration of Sabin IPV and thereby overcome logistic issues currently constraining poliovirus eradication campaigns.
Collapse
Affiliation(s)
- Agnese Donadei
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| | - Heleen Kraan
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands.
| | - Olga Ophorst
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Olivia Flynn
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Conor O'Mahony
- Tyndall National Institute, University College Cork, Cork, Ireland
| | - Peter C Soema
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Anne C Moore
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| |
Collapse
|
28
|
Conjugation of a peptide autoantigen to gold nanoparticles for intradermally administered antigen specific immunotherapy. Int J Pharm 2019; 562:303-312. [DOI: 10.1016/j.ijpharm.2019.03.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/06/2019] [Accepted: 03/18/2019] [Indexed: 01/11/2023]
|
29
|
Effect of HIV-exposure and timing of anti-retroviral treatment on immunogenicity of trivalent live-attenuated polio vaccine in infants. PLoS One 2019; 14:e0215079. [PMID: 31002702 PMCID: PMC6474646 DOI: 10.1371/journal.pone.0215079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/26/2019] [Indexed: 11/30/2022] Open
Abstract
Introduction The prevalence of HIV infection in South African pregnant women has been approximately 30% over the past decade; however, there has been a steady decline in mother-to-child transmission of HIV from 8% in 2008 to <2% in 2015. We evaluated the immunogenicity of live-attenuated trivalent oral polio vaccine (OPV) following the primary vaccination series (doses at birth, 6, 10 and 14 weeks of age) in HIV-exposed uninfected (HEU), HIV-infected infants initiated on early anti-retroviral treatment (HIV+/ART+), HIV-infected infants on deferred ART (HIV+/ART-) and HIV-unexposed infants (HU) as the referent group. Methods Serum polio neutralization antibody titres were evaluated to serotype-1, serotype-2 and serotype-3 at 6, 10 and 18 weeks of age. Antibody titres ≥8 were considered seropositive and sero-protective. Results At 18 weeks of age, following the complete primary series of four OPV doses, no differences in GMTs, percentage of infants with sero-protective titres and median fold change in antibody titre (18 weeks vs 6 weeks) were observed in HEU infants (n = 114) and HIV+/ART+ infants (n = 162) compared to HU infants (n = 104) for the three polio serotypes. However, comparing HIV+/ART- infants (n = 70) to HU infants at 18 weeks of age, we observed significantly lower GMTs for serotype-1 (p = 0.022), serotype-2 (p<0.001) and serotype-3 (p<0.001), significantly lower percentages of infants with sero-protective titres for the three serotypes (p<0.001), and significantly lower median fold change in antibody titre for serotype-1 (p = 0.048), serotype-2 (p = 0.003) and serotype-3 (p = 0.008). Conclusion Delaying initiation of ART in HIV-infected infants was associated with an attenuated immune response to OPV following a four-dose primary series of vaccines, whereas immune responses to OPV in HIV-infected children initiated on ART early in infancy and HEU children were similar to HU infants.
Collapse
|
30
|
Schaumburg F, De Pijper CA, Grobusch MP. Intradermal travel vaccinations-when less means more. Travel Med Infect Dis 2019; 28:3-5. [PMID: 30878310 DOI: 10.1016/j.tmaid.2019.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Münster, Domagkstraße 10, 48149, Münster, Germany.
| | - Cornelis A De Pijper
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection & Immunity, Amsterdam Public Health, Meibergdreef 9, 1100, DD, Amsterdam, Netherlands
| | - Martin P Grobusch
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection & Immunity, Amsterdam Public Health, Meibergdreef 9, 1100, DD, Amsterdam, Netherlands
| |
Collapse
|
31
|
Duarah S, Sharma M, Wen J. Recent advances in microneedle-based drug delivery: Special emphasis on its use in paediatric population. Eur J Pharm Biopharm 2019; 136:48-69. [DOI: 10.1016/j.ejpb.2019.01.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022]
|
32
|
Balasubramanian S, Shah A, Pemde HK, Chatterjee P, Shivananda S, Guduru VK, Soans S, Shastri D, Kumar R. Indian Academy of Pediatrics (IAP) Advisory Committee on Vaccines and Immunization Practices (ACVIP) Recommended Immunization Schedule (2018-19) and Update on Immunization for Children Aged 0 Through 18 Years. Indian Pediatr 2018. [DOI: 10.1007/s13312-018-1444-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
33
|
Cahill EM, Keaveney S, Stuettgen V, Eberts P, Ramos-Luna P, Zhang N, Dangol M, O'Cearbhaill ED. Metallic microneedles with interconnected porosity: A scalable platform for biosensing and drug delivery. Acta Biomater 2018; 80:401-411. [PMID: 30201432 DOI: 10.1016/j.actbio.2018.09.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/01/2018] [Accepted: 09/06/2018] [Indexed: 02/01/2023]
Abstract
Metallic-based microneedles (MNs) offer a robust platform for minimally invasive drug delivery and biosensing applications due to their mechanical strength and proven tissue and drug compatibility. However, current designs suffer from limited functional surface area or challenges in manufacturing scalability. Here, porous 316L stainless steel MN patches are proposed. Fabricated through a scalable manufacturing process, they are suitable for storage and delivery of drugs and rapid absorption of fluids for biosensing. Fabrication of these MNs involves hot embossing a patch of stainless steel-based feedstock, sintering at 1100 °C and subsequent electropolishing. Optimisation of this manufacturing process yields devices that maintain mechanical integrity yet possess high surface area and associated porosity (36%) to maximise loading capacity. Similarly, a small pore size has been targeted (average diameter 2.22 μm, with 90% between 1.56 μm and 2.93 μm) to maximise capillarity and loading efficiency. This porous network has a theoretical wicking rate of 4.7 μl/s and can wick-up 27 ± 5 μl of fluid through capillary action which allows for absorption of pharmaceuticals for delivery. When inserted into a metabolite-loaded skin model, the MNs absorbed and recovered 17 ± 3 μl of the metabolite solution. The drug delivery performance of the porous metallic MNs (22.4 ± 4.9 µg/cm2) was found to be threefold higher than that of topical administration (7.1 ± 4.3 µg/cm2). The porous metallic MN patches have been shown to insert into porcine skin under a 19 N load. These results indicate the potential of design-for-manufacturing porous stainless steel MNs in biosensing and drug delivery applications. STATEMENT OF SIGNIFICANCE: Microneedles are micro-scale sharp protrusions used to bypass the stratum corneum, the skin's outer protective layer, and painlessly access dermal layers suitable for drug delivery and biosensing. Despite a depth of research in the area we have not yet seen large-scale clinical adoption of microneedle devices. Here we describe a device designed to address the potential barriers to adoption seen by other microneedles devices. We have developed a scalable, cost effective process to produce medical grade stainless steel microneedle patches which passively absorb and store drugs or interstitial fluid though a porous network and capillary action. This device, with low manufacturing and regulatory burdens may help the large-scale adoption of microneedles.
Collapse
Affiliation(s)
- Ellen M Cahill
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Shane Keaveney
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Vivien Stuettgen
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Paulina Eberts
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland; Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Pamela Ramos-Luna
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - Nan Zhang
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Manita Dangol
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Eoin D O'Cearbhaill
- UCD Centre for Biomedical Engineering, School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
34
|
Ogai N, Nonaka I, Toda Y, Ono T, Minegishi S, Inou A, Hachiya M, Fukamizu H. Enhanced immunity in intradermal vaccination by novel hollow microneedles. Skin Res Technol 2018; 24:630-635. [PMID: 29707828 DOI: 10.1111/srt.12576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The intradermal (ID) route for vaccination represents an effective alternative to subcutaneous (SC)/intramuscular administration to induce protective immunity. However, a critical issue associated with ID vaccination is the precise delivery of solution in the upper dermis, which ensures enhanced immunity. METHODS We fabricated a hollow microneedle unit made of poly-glycolic acid by injection molding and bonding, and created a dedicated prototype injector. To ensure ID delivery of solution, the injected site was macroscopically and microscopically examined. Serum immunoglobulin G antibody production was measured by enzyme immunoassay and compared in groups of rats following either ID delivery with microneedles or SC administration with a 27-G stainless needle of graded vaccine doses. RESULTS The unit used a tandem array of six microneedles, each with a side delivery hole, and a conduit inside for solution. Microneedles installed in the injector punctured the skin with the aid of a spring. Injection of solution formed a wheal due to ID distribution. Histologically, a wedge-shaped skin defect in the upper skin corresponded to each puncture site. Antibody titers following vaccinations on days 1 and 8 were significantly higher with ID injection than with SC delivery on day 15 and every 7 days thereafter until day 36 with mumps vaccination, and until day 36 with varicella vaccination. CONCLUSIONS The microneedle unit presented here delivered solution intradermally without any difficulty and evoked antibody responses against viruses even with the reduced vaccine volume. Our findings confirm promising results of ID delivery as an immunogenic option to enhance vaccination efficacy.
Collapse
Affiliation(s)
- N Ogai
- ASTI Corporation, Hamamatsu, Japan
| | - I Nonaka
- ASTI Corporation, Hamamatsu, Japan
| | - Y Toda
- ASTI Corporation, Hamamatsu, Japan
| | - T Ono
- ASTI Corporation, Hamamatsu, Japan
| | | | - A Inou
- ASTI Corporation, Hamamatsu, Japan
| | | | - H Fukamizu
- Department of Plastic and Reconstructive Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
35
|
Golombek S, Pilz M, Steinle H, Kochba E, Levin Y, Lunter D, Schlensak C, Wendel HP, Avci-Adali M. Intradermal Delivery of Synthetic mRNA Using Hollow Microneedles for Efficient and Rapid Production of Exogenous Proteins in Skin. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:382-392. [PMID: 29858073 PMCID: PMC5992458 DOI: 10.1016/j.omtn.2018.03.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/21/2022]
Abstract
In recent years, synthetic mRNA-based applications to produce desired exogenous proteins in cells have been gaining importance. However, systemic delivery of synthetic mRNA can result in unspecific uptake into undesired cells or organs and, thereby, fail to target desired cells. Thus, local and targeted delivery of synthetic mRNA becomes increasingly important to reach the desired cell types and tissues. In this study, intradermal delivery of synthetic mRNA using a hollow microneedle injection-based method was evaluated. Furthermore, an ex vivo porcine skin model was established to analyze synthetic mRNA-mediated protein expression in the skin following intradermal delivery. Using this model, highly efficient delivery of synthetic mRNA was demonstrated, which resulted in detection of high levels of secretable humanized Gaussia luciferase (hGLuc) protein encoded by the microinjected synthetic mRNA. Interestingly, synthetic mRNA injected without transfection reagent was also able to enter the cells and resulted in protein expression. The established ex vivo porcine skin model can be used to evaluate the successful production of desired proteins after intradermal delivery of synthetic mRNAs before starting with in vivo experiments. Furthermore, the use of microneedles enables patient-friendly, painless, and efficient delivery of synthetic mRNAs into the dermis; thus, this method could be applied for local treatment of different skin diseases as well as for vaccination and immunotherapy.
Collapse
Affiliation(s)
- Sonia Golombek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Martin Pilz
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Heidrun Steinle
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Efrat Kochba
- NanoPass Technologies Ltd., 3 Golda Meir, 7403648 Nes Ziona, Israel
| | - Yotam Levin
- NanoPass Technologies Ltd., 3 Golda Meir, 7403648 Nes Ziona, Israel
| | - Dominique Lunter
- Department of Pharmaceutical Technology, Eberhard Karls University, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| |
Collapse
|
36
|
Microneedles as the technique of drug delivery enhancement in diverse organs and tissues. J Control Release 2018; 270:184-202. [DOI: 10.1016/j.jconrel.2017.11.048] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 11/24/2022]
|
37
|
|
38
|
Muller DA, Fernando GJP, Owens NS, Agyei-Yeboah C, Wei JCJ, Depelsenaire ACI, Forster A, Fahey P, Weldon WC, Oberste MS, Young PR, Kendall MAF. High-density microprojection array delivery to rat skin of low doses of trivalent inactivated poliovirus vaccine elicits potent neutralising antibody responses. Sci Rep 2017; 7:12644. [PMID: 28974777 PMCID: PMC5626768 DOI: 10.1038/s41598-017-13011-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/12/2017] [Indexed: 01/09/2023] Open
Abstract
To secure a polio-free world, the live attenuated oral poliovirus vaccine (OPV) will eventually need to be replaced with inactivated poliovirus vaccines (IPV). However, current IPV delivery is less suitable for campaign use than OPV, and more expensive. We are progressing a microarray patch delivery platform, the Nanopatch, as an easy-to-use device to administer vaccines, including IPV. The Nanopatch contains an ultra-high density array (10,000/cm2) of short (~230 μm) microprojections that delivers dry coated vaccine into the skin. Here, we compare the relative immunogenicity of Nanopatch immunisation versus intramuscular injection in rats, using monovalent and trivalent formulations of IPV. Nanopatch delivery elicits faster antibody response kinetics, with high titres of neutralising antibody after just one (IPV2) or two (IPV1 and IPV3) immunisations, while IM injection requires two (IPV2) or three (IPV1 and IPV3) immunisations to induce similar responses. Seroconversion to each poliovirus type was seen in 100% of rats that received ~1/40th of a human dose of IPV delivered by Nanopatch, but not in rats given ~1/8th or ~1/40th dose by IM injection. Ease of administration coupled with dose reduction observed in this study suggests the Nanopatch could facilitate inexpensive IPV vaccination in campaign settings.
Collapse
Affiliation(s)
- David A Muller
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia. .,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia. .,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia.
| | - Germain J P Fernando
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland, Australia
| | - Nick S Owens
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia
| | - Christiana Agyei-Yeboah
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia
| | - Jonathan C J Wei
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia
| | - Alexandra C I Depelsenaire
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia
| | - Angus Forster
- Vaxxas Pty Ltd, Translational Research Institute, Brisbane, Queensland, 4102, Australia
| | - Paul Fahey
- Vaxxas Pty Ltd, Translational Research Institute, Brisbane, Queensland, 4102, Australia
| | - William C Weldon
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - M Steven Oberste
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Paul R Young
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark A F Kendall
- Delivery of Drugs and Genes Group (D2G2) Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, QLD 4072, Australia. .,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland, Australia.
| |
Collapse
|
39
|
Crum-Cianflone NF, Sullivan E. Vaccinations for the HIV-Infected Adult: A Review of the Current Recommendations, Part II. Infect Dis Ther 2017; 6:333-361. [PMID: 28780736 PMCID: PMC5595779 DOI: 10.1007/s40121-017-0165-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Indexed: 12/14/2022] Open
Abstract
Vaccination is a critical component for ensuring the ongoing health HIV-infected adults. Since this group may have reduced immune responses and shorter durations of protection post-vaccination, HIV-specific guidelines have been published. This review article provides a comprehensive discussion of the current guidelines and evidence-based data for vaccinating HIV-infected adults, including data on dosing schedules, immunogenicity studies, and safety. In the current paper, part II of the review, live vaccines, as well as vaccines for travelers and specific occupational groups, will be discussed.
Collapse
Affiliation(s)
- Nancy F Crum-Cianflone
- Internal Medicine Department, Scripps Mercy Hospital, San Diego, CA, USA.
- Infectious Disease Division, Scripps Mercy Hospital, San Diego, CA, USA.
- Infectious Disease Division, Naval Medical Center San Diego, San Diego, CA, USA.
| | - Eva Sullivan
- Pharmacy Department, Scripps Mercy Hospital, San Diego, CA, USA
| |
Collapse
|
40
|
Bhatnagar S, Dave K, Venuganti VVK. Microneedles in the clinic. J Control Release 2017; 260:164-182. [DOI: 10.1016/j.jconrel.2017.05.029] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/21/2017] [Accepted: 05/23/2017] [Indexed: 12/16/2022]
|
41
|
Okayasu H, Sein C, Chang Blanc D, Gonzalez AR, Zehrung D, Jarrahian C, Macklin G, Sutter RW. Intradermal Administration of Fractional Doses of Inactivated Poliovirus Vaccine: A Dose-Sparing Option for Polio Immunization. J Infect Dis 2017; 216:S161-S167. [PMID: 28838185 PMCID: PMC5853966 DOI: 10.1093/infdis/jix038] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A fractional dose of inactivated poliovirus vaccine (fIPV) administered by the intradermal route delivers one fifth of the full vaccine dose administered by the intramuscular route and offers a potential dose-sparing strategy to stretch the limited global IPV supply while further improving population immunity. Multiple studies have assessed immunogenicity of intradermal fIPV compared with the full intramuscular dose and demonstrated encouraging results. Novel intradermal devices, including intradermal adapters and disposable-syringe jet injectors, have also been developed and evaluated as alternatives to traditional Bacillus Calmette-Guérin needles and syringes for the administration of fIPV. Initial experience in India, Pakistan, and Sri Lanka suggests that it is operationally feasible to implement fIPV vaccination on a large scale. Given the available scientific data and operational feasibility shown in early-adopter countries, countries are encouraged to consider introducing a fIPV strategy into their routine immunization and supplementary immunization activities.
Collapse
MESH Headings
- Antibodies, Viral/immunology
- Child
- Child, Preschool
- Humans
- Immunization, Secondary/economics
- Immunization, Secondary/methods
- Infant
- Injections, Intradermal/instrumentation
- Injections, Intradermal/methods
- Mass Vaccination/economics
- Mass Vaccination/instrumentation
- Mass Vaccination/methods
- Poliovirus/immunology
- Poliovirus Vaccine, Inactivated/administration & dosage
- Poliovirus Vaccine, Inactivated/economics
- Poliovirus Vaccine, Inactivated/immunology
- Poliovirus Vaccine, Inactivated/supply & distribution
Collapse
Affiliation(s)
| | - Carolyn Sein
- Research, Policy and Containment, Polio Eradication Department
| | - Diana Chang Blanc
- Expanded Programme on Immunization, Immunization, Vaccines and Biologicals Department, World Health Organization, Geneva, Switzerland
| | - Alejandro Ramirez Gonzalez
- Expanded Programme on Immunization, Immunization, Vaccines and Biologicals Department, World Health Organization, Geneva, Switzerland
| | | | | | - Grace Macklin
- Research, Policy and Containment, Polio Eradication Department
| | - Roland W Sutter
- Research, Policy and Containment, Polio Eradication Department
| |
Collapse
|
42
|
Brief Report: Seroprevalence of Pertussis Infection in HIV-Infected Adults in the United States. J Acquir Immune Defic Syndr 2017; 73:282-286. [PMID: 27105050 DOI: 10.1097/qai.0000000000001037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pertussis is a resurgent infection that can cause significant morbidity among adults. CD4 T cells are necessary for its clearance, but pertussis studies in HIV-infected adults are limited to case reports. We analyzed stored serum samples from 299 HIV-infected adults to determine the seroprevalence of pertussis among this population. We found that 4.3% of subjects had serologic evidence of recent pertussis infection, and annual incidence of pertussis infection among subjects not vaccinated against pertussis in the last 5 years was 10.5%-17.5%. Prospective studies are needed to define the clinical presentation of pertussis in HIV-infected adults and to optimize vaccination strategies.
Collapse
|
43
|
Effects of Well-Controlled HIV Infection on Complement Activation and Function. J Acquir Immune Defic Syndr 2017; 73:20-6. [PMID: 27192377 DOI: 10.1097/qai.0000000000001079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Uncontrolled HIV infection is known to activate the complement system, leading to an increase in chronic inflammation. Whether or not this activation of complement persists and contributes to chronic inflammation in subjects with HIV infection that is well controlled through use of antiretroviral therapy has not been studied. METHODS We conducted an observational, cross-sectional study using sera from 305 adults with well-controlled HIV infection and 30 healthy controls. Sera was tested for markers of complement activation (C3a and C5a levels), complement function (CH50 assay), and immunoglobulin levels (IgG1-IgG4) as IgG can activate complement. We evaluated the association of well-controlled HIV infection with C3a, C5a, CH50, IgG1-IgG4, and total IgG levels using both univariate and multivariate analyses, controlling for factors such as age, sex, race, comorbidities (including hepatitis C coinfection), smoking status, and statin use. RESULTS Well-controlled HIV infection was associated with a 54% increase in complement activation as measured by C3a levels compared with healthy controls (P < 0.0001). Hepatitis C coinfection was associated with a further 52% increase in complement activation, as measured by C3a levels, over HIV alone (P = 0.003). CONCLUSION These results suggest that complement activation may contribute to a proinflammatory state even in well-controlled HIV infection. Furthermore, hepatitis C virus coinfection may be even more proinflammatory, in complement activation, compared with HIV infection alone.
Collapse
|
44
|
Jarrahian C, Rein-Weston A, Saxon G, Creelman B, Kachmarik G, Anand A, Zehrung D. Vial usage, device dead space, vaccine wastage, and dose accuracy of intradermal delivery devices for inactivated poliovirus vaccine (IPV). Vaccine 2017; 35:1789-1796. [PMID: 28189403 PMCID: PMC6381447 DOI: 10.1016/j.vaccine.2016.11.098] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/15/2016] [Accepted: 11/29/2016] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Intradermal delivery of a fractional dose of inactivated poliovirus vaccine (IPV) offers potential benefits compared to intramuscular (IM) delivery, including possible cost reductions and easing of IPV supply shortages. Objectives of this study were to assess intradermal delivery devices for dead space, wastage generated by the filling process, dose accuracy, and total number of doses that can be delivered per vial. METHODS Devices tested included syringes with staked (fixed) needles (autodisable syringes and syringes used with intradermal adapters), a luer-slip needle and syringe, a mini-needle syringe, a hollow microneedle device, and disposable-syringe jet injectors with their associated filling adapters. Each device was used to withdraw 0.1-mL fractional doses from single-dose IM glass vials which were then ejected into a beaker. Both vial and device were weighed before and after filling and again after expulsion of liquid to record change in volume at each stage of the process. Data were used to calculate the number of doses that could potentially be obtained from multidose vials. RESULTS Results show wide variability in dead space, dose accuracy, overall wastage, and total number of doses that can be obtained per vial among intradermal delivery devices. Syringes with staked needles had relatively low dead space and low overall wastage, and could achieve a greater number of doses per vial compared to syringes with a detachable luer-slip needle. Of the disposable-syringe jet injectors tested, one was comparable to syringes with staked needles. DISCUSSION If intradermal delivery of IPV is introduced, selection of an intradermal delivery device can have a substantial impact on vaccine wasted during administration, and thus on the required quantity of vaccine that needs to be purchased. An ideal intradermal delivery device should be not only safe, reliable, accurate, and acceptable to users and vaccine recipients, but should also have low dead space, high dose accuracy, and low overall wastage to maximize the potential number of doses that can be withdrawn and delivered.
Collapse
Affiliation(s)
| | | | - Gene Saxon
- PATH, PO Box 900922, Seattle, WA 98109, USA
| | | | | | - Abhijeet Anand
- Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30333, USA
| | | |
Collapse
|
45
|
Lal M, Jarrahian C. Presentation matters: Buffers, packaging, and delivery devices for new, oral enteric vaccines for infants. Hum Vaccin Immunother 2017; 13:46-49. [PMID: 27819524 PMCID: PMC5287318 DOI: 10.1080/21645515.2016.1238536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/14/2016] [Indexed: 11/01/2022] Open
Abstract
Oral administration of vaccines is simpler and more acceptable than injection via needle and syringe, particularly for infants (Fig. 1) This route is promising for new vaccines in development against enterotoxigenic Escherichia coli (ETEC) and Shigella that cause childhood diarrhea with devastating consequences in low-resource countries. However, vaccine antigens and adjuvants given orally need buffering against the degradative effects of low stomach pH, and the type and volume of antacid buffer require special attention for infants. In addition, container/closure systems must be compatible with vaccine formulations, protect against water and gas transfer, and have minimal impact on the cold chain. Health care workers in demanding low-resource settings need an administration device that is easy to use, yet will accurately measure and safely deliver the correct vaccine dose. Developers must consider manufacturing capabilities, and immunization program managers want affordable vaccines. As new combination enteric vaccine candidates advance into clinical evaluation, features of the final vaccine presentation-liquid or dry format, diluent, buffer, primary and secondary packaging, and administration device-should be taken into account early in product development to achieve the greatest possible impact for the vaccine.
Collapse
|
46
|
Seo YB, Lee J, Song JY, Choi HJ, Cheong HJ, Kim WJ. Safety and immunogenicity of influenza vaccine among HIV-infected adults: Conventional vaccine vs. intradermal vaccine. Hum Vaccin Immunother 2016; 12:478-84. [PMID: 26431466 DOI: 10.1080/21645515.2015.1076599] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Several studies have reported poor immune responses to conventional influenza vaccines in HIV-infected individuals. This study sought to elicit more potent immunogenicity in HIV-infected adults using an intradermal vaccine compared with a conventional intramuscular vaccine. This multicenter, randomized, controlled, open-label study was conducted at 3 university hospitals during the 2011/2012 pre-influenza season. Three vaccines were used in HIV-infected adults aged 18 - 60 years: an inactivated intramuscular vaccine (Agrippal), a reduced-content intradermal vaccine (IDflu9μg) and a standard-content intradermal vaccine (IDflu15μg). Serum hemagglutination-inhibiting (HI) antibodies and INF-γ ELISpot assay were measured at the time of vaccination and 1 month after vaccination. Adverse events were recorded for 7 d. A total of 28 Agrippal, 30 IDflu9μg, and 28 IDflu15μg volunteers were included in this analysis. One month after vaccination, the GMTs and differences in INF-γ ELISpot assay results were similar among the 3 groups. Seroprotection rates, seroconversion rates and mean fold increases (MFI) among the 3 groups were also similar, at approximately 80%, 50-60% and 2.5 - 10.0, respectively. All three vaccines satisfied the CHMP criteria for the A/H1N1 and A/H3N2 strains, but not those for the B strain. In univariate analysis, no demographic or clinical factors, including age, CD4+ T-cell counts, HIV viral load, ART status and vaccine type, were related to failure to achieve seroprotection. The three vaccines were all well-tolerated and all reported reactions were mild to moderate. However, there was a tendency toward a higher incidence of local and systemic reactions in the intradermal vaccine groups. The intradermal vaccine did not result in higher immunogenicity compared to the conventional intramuscular vaccine, even with increased antigen dose.
Collapse
Affiliation(s)
- Yu Bin Seo
- a Division of Infectious Diseases ; Department of Internal Medicine; Hallym University College of Medicine ; Chuncheon , Republic of Korea
| | - Jacob Lee
- a Division of Infectious Diseases ; Department of Internal Medicine; Hallym University College of Medicine ; Chuncheon , Republic of Korea
| | - Joon Young Song
- b Division of Infectious Diseases ; Department of Internal Medicine; Korea University College of Medicine ; Seoul , Republic of Korea
| | - Hee Jung Choi
- c Division of Infectious Diseases ; Department of Internal Medicine; Ewha Woman University College of Medicine ; Seoul , Republic of Korea
| | - Hee Jin Cheong
- b Division of Infectious Diseases ; Department of Internal Medicine; Korea University College of Medicine ; Seoul , Republic of Korea
| | - Woo Joo Kim
- b Division of Infectious Diseases ; Department of Internal Medicine; Korea University College of Medicine ; Seoul , Republic of Korea
| |
Collapse
|
47
|
Wang Y, Vlasova A, Velasquez DE, Saif LJ, Kandasamy S, Kochba E, Levin Y, Jiang B. Skin Vaccination against Rotavirus Using Microneedles: Proof of Concept in Gnotobiotic Piglets. PLoS One 2016; 11:e0166038. [PMID: 27824918 PMCID: PMC5100943 DOI: 10.1371/journal.pone.0166038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 10/22/2016] [Indexed: 01/03/2023] Open
Abstract
Live-attenuated oral rotavirus (RV) vaccines have lower efficacy in low income countries, and additionally are associated with a rare but severe adverse event, intussusception. We have been pursuing the development of an inactivated rotavirus vaccine (IRV) using the human rotavirus strain CDC-9 (G1P[8]) through parenteral immunization and previously demonstrated dose sparing and enhanced immunogenicity of intradermal (ID) unadjuvanted IRV using a coated microneedle patch in comparison with intramuscular (IM) administration in mice. The aim of this study was to evaluate the immune response and protection against RV infection and diarrhea conferred by the administration of the ID unadjuvanted IRV using the microneedle device MicronJet600® in neonatal gnotobiotic (Gn) piglets challenged with virulent Wa G1P[8] human RV. Three doses of 5 μg IRV when administered intradermally and 5 μg IRV formulated with aluminum hydroxide [Al(OH)3] when administered intramuscularly induced comparable rotavirus-specific antibody titers of IgA, IgG, IgG avidity index and neutralizing activity in sera of neonatal piglets. Both IRV vaccination regimens protected against RV antigen shedding in stools, and reduced the cumulative diarrhea scores in the piglets. This study demonstrated that the ID and IM administrations of IRV are immunogenic and protective against RV-induced diarrhea in neonatal piglets. Our findings highlight the potential value of an adjuvant sparing effect of the IRV ID delivery route.
Collapse
Affiliation(s)
- Yuhuan Wang
- Gastroenteritis and Respiratory Viruses Laboratory Branch Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Anastasia Vlasova
- Food Animal Health Research Program, Ohio Agricultural Research & Development Center, The Ohio State University, Wooster, Ohio, United States of America
| | - Daniel E. Velasquez
- Gastroenteritis and Respiratory Viruses Laboratory Branch Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Linda J. Saif
- Food Animal Health Research Program, Ohio Agricultural Research & Development Center, The Ohio State University, Wooster, Ohio, United States of America
| | - Sukumar Kandasamy
- Food Animal Health Research Program, Ohio Agricultural Research & Development Center, The Ohio State University, Wooster, Ohio, United States of America
| | | | - Yotam Levin
- NanoPass Technologies Ltd., Nes Ziona, Israel
| | - Baoming Jiang
- Gastroenteritis and Respiratory Viruses Laboratory Branch Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
48
|
Schipper P, van der Maaden K, Romeijn S, Oomens C, Kersten G, Jiskoot W, Bouwstra J. Repeated fractional intradermal dosing of an inactivated polio vaccine by a single hollow microneedle leads to superior immune responses. J Control Release 2016; 242:141-147. [PMID: 27496634 DOI: 10.1016/j.jconrel.2016.07.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/26/2016] [Accepted: 07/31/2016] [Indexed: 12/28/2022]
Abstract
The purpose of this study was to investigate the effect of various repeated fractional intradermal dosing schedules of inactivated polio vaccine serotype 1 (IPV1) on IPV1-specific IgG responses in rats. By utilizing an applicator that allowed for precisely controlled intradermal microinjections by using a single hollow microneedle, rats were immunized intradermally with 5 D-antigen units (DU) of IPV1 at 150μm skin depth. This dose was administered as a bolus, or in a repeated fractional dosing schedule: 4 doses of 1.25 DU (1/4th of total dose) were administered on four consecutive days or every other day; 8 doses of 0.625 DU (1/8th of total dose) were administered on eight consecutive days; or 4 exponentially increasing doses (0.04, 0.16, 0.8 and 4 DU), either with or without an exponentially increasing CpG oligodeoxynucleotide 1826 (CpG) dose, were administered on four consecutive days. All of these fractional dosing schedules resulted in up to ca. 10-fold higher IPV1-specific IgG responses than intradermal and intramuscular bolus dosing. IPV1 combined with adjuvant CpG in exponential dosing did not significantly increase the IPV1-specific IgG responses further, which demonstrated that maximal responses were achieved by fractional dosing. In conclusion, repeated fractional intradermal IPV1 dosing leads to superior IPV1-specific IgG responses without the use of adjuvants. These results indicate that a controlled release delivery system for intradermal IPV1 delivery can potentiate IPV1-specific IgG responses.
Collapse
Affiliation(s)
- Pim Schipper
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Koen van der Maaden
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Stefan Romeijn
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Cees Oomens
- Soft Tissue Biomechanics and Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Gideon Kersten
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands.
| | - Wim Jiskoot
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Joke Bouwstra
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
49
|
Schipper P, van der Maaden K, Romeijn S, Oomens C, Kersten G, Jiskoot W, Bouwstra J. Determination of Depth-Dependent Intradermal Immunogenicity of Adjuvanted Inactivated Polio Vaccine Delivered by Microinjections via Hollow Microneedles. Pharm Res 2016; 33:2269-79. [PMID: 27317570 DOI: 10.1007/s11095-016-1965-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/27/2016] [Indexed: 01/30/2023]
Abstract
PURPOSE The aim of this study was to investigate the depth-dependent intradermal immunogenicity of inactivated polio vaccine (IPV) delivered by depth-controlled microinjections via hollow microneedles (HMN) and to investigate antibody response enhancing effects of IPV immunization adjuvanted with CpG oligodeoxynucleotide 1826 (CpG) or cholera toxin (CT). METHODS A novel applicator for HMN was designed to permit depth- and volume-controlled microinjections. The applicator was used to immunize rats intradermally with monovalent IPV serotype 1 (IPV1) at injection depths ranging from 50 to 550 μm, or at 400 μm for CpG and CT adjuvanted immunization, which were compared to intramuscular immunization. RESULTS The applicator allowed accurate microinjections into rat skin at predetermined injection depths (50-900 μm), -volumes (1-100 μL) and -rates (up to 60 μL/min) with minimal volume loss (±1-2%). HMN-mediated intradermal immunization resulted in similar IgG and virus-neutralizing antibody titers as conventional intramuscular immunization. No differences in IgG titers were observed as function of injection depth, however IgG titers were significantly increased in the CpG and CT adjuvanted groups (7-fold). CONCLUSION Intradermal immunogenicity of IPV1 was not affected by injection depth. CpG and CT were potent adjuvants for both intradermal and intramuscular immunization, allowing effective vaccination upon a minimally-invasive single intradermal microinjection by HMN.
Collapse
Affiliation(s)
- Pim Schipper
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Koen van der Maaden
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Stefan Romeijn
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Cees Oomens
- Soft Tissue Biomechanics and Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Gideon Kersten
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Wim Jiskoot
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Joke Bouwstra
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands.
| |
Collapse
|
50
|
Abstract
Intradermal (ID) vaccination induces a more potent immune response and requires lower vaccine doses as compared with standard vaccination routes. To deliver ID vaccines effectively and consistently, an ID delivery device has been developed and is commercially available for adults. The clinical application of ID vaccines for infants and children is much anticipated because children receive several vaccines, on multiple occasions, during infancy and childhood. However, experience with ID vaccines is limited and present evidence is sparse and inconsistent. ID delivery devices are not currently available for infants and children, but recent studies have examined skin thickness in this population and reported that it did not differ in proportion to body size in infants, children, and adults. These results are helpful in developing new ID devices and for preparing new vaccines in infants and children.
Collapse
Affiliation(s)
- Akihiko Saitoh
- a Department of Pediatrics , Niigata University Graduate School of Medical and Dental Sciences , Niigata , Japan
| | - Yuta Aizawa
- a Department of Pediatrics , Niigata University Graduate School of Medical and Dental Sciences , Niigata , Japan
| |
Collapse
|